Anda di halaman 1dari 11

Molecular and Cellular Endocrinology 361 (2012) 111

Contents lists available at SciVerse ScienceDirect

Molecular and Cellular Endocrinology


journal homepage: www.elsevier.com/locate/mce

Review

Hepatic growth hormone and glucocorticoid receptor signaling in body


growth, steatosis and metabolic liver cancer development
Kristina M. Mueller a, Madeleine Themanns a, Katrin Friedbichler a, Jan-Wilhelm Kornfeld b,
Harald Esterbauer c, Jan P. Tuckermann d,e, Richard Moriggl a,
a
Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
b
Institute for Genetics, Department of Mouse Genetics and Metabolism, University of Cologne, Cologne, Germany
c
Department of Laboratory Medicine, Medical University Vienna, Vienna, Austria
d
Tissue-Specic Hormone Action, Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany
e
Institute for General Zoology and Endocrinology, University of Ulm, Ulm, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Growth hormone (GH) and glucocorticoids (GCs) are involved in the control of processes that are essen-
Received 28 February 2012 tial for the maintenance of vital body functions including energy supply and growth control. GH and GCs
Accepted 30 March 2012 have been well characterized to regulate systemic energy homeostasis, particular during certain condi-
Available online 30 April 2012
tions of physical stress. However, dysfunctional signaling in both pathways is linked to various metabolic
disorders associated with aberrant carbohydrate and lipid metabolism. In liver, GH-dependent activation
Keywords: of the transcription factor signal transducer and activator of transcription (STAT) 5 controls a variety of
STAT5
physiologic functions within hepatocytes. Similarly, GCs, through activation of the glucocorticoid recep-
Non-alcoholic fatty liver disease
Glucose metabolism
tor (GR), inuence many important liver functions such as gluconeogenesis. Studies in hepatic Stat5 or GR
Glucocorticoids knockout mice have revealed that they similarly control liver function on their target gene level and
Growth hormone indeed, the GR functions often as a cofactor of STAT5 for GH-induced genes. Gene sets, which require
physical STAT5GR interaction, include those controlling body growth and maturation. More recently,
it has become evident that impairment of GH-STAT5 signaling in different experimental models corre-
lates with metabolic liver disease, ranging from hepatic steatosis to hepatocellular carcinoma (HCC).
While GH-activated STAT5 has a protective role in chronic liver disease, experimental disruption of
GC-GR signaling rather seems to ameliorate metabolic disorders under metabolic challenge. In this
review, we focus on the current knowledge about hepatic GH-STAT5 and GC-GR signaling in body growth,
metabolism, and protection from fatty liver disease and HCC development.
2012 Elsevier Ireland Ltd. Open access under CC BY-NC-ND license.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Mechanism of GH-STAT5 and GC-GR action. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. GH signaling and STAT5 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. GC signaling and GR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.3. STAT5 and GR synergism in hepatocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3. STAT5 and GR function in body growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
4. GH-STAT5 and GR function in metabolism and hepatic steatosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
4.1. Glucose metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
4.2. Lipid metabolism and hepatic steatosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5. GH-STAT5 signaling and HCC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
6. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9

Corresponding author. Address: Ludwig Boltzmann Institute for Cancer Research, Waehringerstrasse 13a, 1090 Vienna, Austria. Tel.: +43 14277 64111; fax: +43 14277 9641.
E-mail address: richard.moriggl@lbicr.lbg.ac.at (R. Moriggl).

0303-7207 2012 Elsevier Ireland Ltd. Open access under CC BY-NC-ND license.
http://dx.doi.org/10.1016/j.mce.2012.03.026
2 K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111

1. Introduction strict hormonal control. Hypothalamic growth hormone-releasing


hormone (GHRH) is the central stimulator of GH synthesis, while
The liver plays an important role in whole-body metabolism hypothalamic somatostatin exerts strong inhibitory effects
and energy homeostasis. Depending on the bodys needs, hepato- (Schneider et al., 2003). Other factors stimulating GH release are
cytes coordinate these processes by regulating gene expression acute stress and energy deprivation (Moller and Jorgensen, 2009),
programs in response to various humoral signals. Imbalances in whereas overnutrition and obesity inhibit its secretion (Scacchi
this control system are associated with a variety of liver patholo- et al., 1999; Flores-Morales et al., 2006).
gies ranging from hepatic steatosis to end-stage liver disease At the cellular level, GH action is mediated via the GH receptor
including hepatocellular carcinoma (HCC) (Feldstein, 2010; (GHR), which is widely expressed in many tissues such as liver,
Bechmann et al., 2011). muscle and adipose tissue. GH signaling is very similar both from
Growth hormone (GH) and glucocorticoids (GCs) are necessary for receptor binding and signal transduction to prolactin, erythropoie-
normal growth and development, as well as immune functions tin and thrombopoietin signaling. These four cytokine signaling
(Sapolsky et al., 2000; Jeay et al., 2002). Further, both factors are receptors all make homodimers upon cytokine binding, and their
important regulators of whole body energy homeostasis (Vegiopoulos main tyrosine kinase responsible for signal transduction is the
and Herzig, 2007; Moller and Jorgensen, 2009; Vijayakumar et al., cytoplasmic Janus kinase 2 (JAK2). GHR binding leads to the activa-
2010). At the cellular level, GH action is mediated amongst others tion of multiple signaling pathways, including the RAS/RAF/ERK,
by the signal transducer and activator of transcription (STAT) 5 the PI3K and the JAK/STAT pathways (Lanning and Carter-Su,
pathway. Through STAT5, GH controls many features of liver phys- 2006; Waters et al., 2006). Although STAT1 and STAT3 can be acti-
iology, including regulation of genes associated with somatic vated through GHR signaling, STAT5 activation is the major target
growth and maturation (Hennighausen and Robinson, 2008). GCs (Zhu et al., 2001) (Fig. 1). Yet, in the absence of STAT5 expression,
exert their functions through cell-specic action of the glucocorti- increased STAT1 and STAT3 activation was reported (Cui et al.,
coid receptor (GR). The GR acts as a transcriptional regulator of dis- 2007; Mueller et al., 2011). STAT5 consists of two different but
tinct target genes via direct DNA binding or through proteinprotein highly homologous isoforms STAT5A and STAT5B (referred to as
interactions with other transcription factors (Kassel and Herrlich, STAT5), which are encoded by two juxtaposed genes on mouse
2007). chromosome 11 and human chromosome 17. Both STAT5 isoforms
Whole-body STAT5- or GR-knockout mice are not viable, dem- differ in their tissue distribution (Hennighausen and Robinson,
onstrating the importance of both transcription factors for develop- 2008). Activated GHR brings two JAK2 molecules into proximity
ment and survival (Cole et al., 1995; Cui et al., 2004). Therefore, which causes auto activation, and subsequent tyrosine phosphory-
conditional knockouts were key to explore the functions of both lation of predimerized STAT5 molecules. Subsequently, activated
transcription factors in liver physiology. Hepatic deciency of STAT5 translocates to the nucleus, where it binds specic DNA
STAT5 or the GR results in a comparable retardation of postnatal binding response elements (REs), usually an inverted repeat of
body growth (Tronche et al., 2004; Engblom et al., 2007). From TTCN3GAA, to modulate target gene transcription (Zhu et al.,
these studies, it became evident that transcription of distinct STAT5 2001; Kornfeld et al., 2008). STAT5 was also shown to be an ef-
target-gene subsets requires binding of the GR onto the STAT5B N- cient chromatin regulator and it can induce loop formation
terminus, the major STAT5 isoform expressed in the liver. This through oligomerization via the N-terminus (Moriggl et al., in
interaction preferentially affects gene sets involved in somatic press; Kornfeld et al., 2008). Whether hepatic STAT5B participates
growth and maturation (Engblom et al., 2007). Yet, although both in oligomerization is controversial, yet, multiple STAT5 REs in tar-
transcription factors are important for the maintenance of energy get genes such as in Socs2 and Igf-1 suggests that possibility (Laz
homeostasis, the phenotypes obtained upon conditional deletion et al., 2009). Amongst other functions, the classical STAT5 target
of either gene in mice with regard to lipid and glucose metabolism genes Igf1 and Socs2 serve to down-regulate GH signaling, thereby
are rather distinct. Interference with hepatic GC-GR signaling is establishing a negative feedback loop (Flores-Morales et al., 2006;
associated with defects in gluconeogenesis leading to fasting hypo- Cui et al., 2007).
glycemia, with no effects on hepatic lipid homeostasis under basal
conditions (Opherk et al., 2004; Mueller et al., 2011). In contrast, 2.2. GC signaling and GR
impairment of hepatic GH-STAT5 signaling is associated with aber-
rant glucose metabolism, fatty liver disease and it sensitizes hepa- Main biological functions of the GC-GR pathway include the
tocytes to injury and tumorigenic transformation (Cui et al., 2007; suppression of inammation (Webster et al., 2002) and the control
Fan et al., 2009; Hosui et al., 2009; Mueller et al., 2011; Friedbichler of energy metabolism in metabolically active organs (Vegiopoulos
et al., 2012). and Herzig, 2007). Secretion of GCs by the adrenal cortex is under
In the following, we will summarize the consequences of genet- control of the hypothalamicpituitaryadrenal (HPA) axis, a neuro-
ic GR and/or Stat5 deletion for body growth, metabolic homeosta- endocrine feedback system. Activation of the HPA axis is inu-
sis, hepatic steatosis and HCC development. enced by various stressors, such as energy deprivation, by the
circadian clock and inammation (Tsigos and Chrousos, 2002;
Buckley and Schatzberg, 2005; Lamia et al., 2011). The subsequent
2. Mechanism of GH-STAT5 and GC-GR action release of hypothalamic corticotropin releasing hormone (CRH)
stimulates synthesis and secretion of pituitary adrenocorticotropic
2.1. GH signaling and STAT5 hormone (ACTH) and the following ACTH-induced stimulation of
adrenal GC synthesis. GCs, in turn, control the regulation of basal
GH is an important regulator of postnatal body growth. In con- HPA axis activity, thereby establishing a regulatory feedback loop.
trast to a nuclear hormone receptor steroid ligand it is a peptide Cellular action of GCs is attributed to their binding to intracel-
and belongs to the superfamily of cytokines. GH controls regenera- lular GR, a member of the nuclear hormone receptor family (Kassel
tion and cellular reproduction. In addition, GH exerts important and Herrlich, 2007; Beck et al., 2011). The inactive GR is retained in
functions in energy metabolism (Moller and Jorgensen, 2009; the cytoplasm complexed with chaperones (e.g. Heat Shock Pro-
Vijayakumar et al., 2010) and inuences the immune system (Jeay tein-90 and Heat Shock Protein-70) until binding to its ligand. After
et al., 2002). GH is part of the somatotropic axis and it is synthesized ligand binding, GR dissociates from the multi-protein complex and
and secreted by the anterior pituitary gland. Its secretion is under either interferes with signal transduction components in the
K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111 3

Fig. 1. Signal transduction pathways induced by GH and GCs. GH binding to a GHR dimer induces a conformational change that activates two JAK2 molecules which results in
phosphorylation of multiple tyrosine residues in the cytoplasmic domain of the GHR. The activated GHR-JAK2 complex facilitates activation of STAT5 proteins by tyrosine
phosphorylation. Activation of STAT5 allows GH to elicit diverse biological and physiological effects. STAT5 targets include anti-apoptotic genes and genes that promote cell
cycle progression as well as inhibition. STAT5 proteins also regulate expression of genes involved in growth, differentiation, RNA biogenesis and metabolism. In addition, GH-
activated JAK2 activates multiple signaling proteins and pathways including STAT1/STAT3, MAPK and PI3K signaling. The binding of GH to GHR may also activate Src tyrosine
kinase, initiating other signaling pathways. The GR is retained in the cytosol as part of a chaperone-containing multiprotein complex. GCs can diffuse freely across the plasma
membrane. Upon ligand binding, GR dissociates from its chaperoning complex and translocates into the nucleus, where it exerts transcriptional effects via direct DNA binding
at GREs or through a GRE-independent distinct proteinprotein interaction mechanism. GR target genes include amongst others rate limiting enzymes of gluconeogenesis.
Additionally, the GR regulates GH-STAT5-dependent transcription of gene sets involved in postnatal body growth and maturation in a cofactor-dependent manner (displayed
in gray). GH, Growth hormone; GCs, glucocorticoids; GHR, GH receptor; JAK2, janus kinase 2; STAT, signal transducer and activator of transcription; SOCS2, suppressor of
cytokine signaling 2; CIS, Cytokine Inducible SH2 containing protein; MAPK, mitogen-activated protein kinase; GR, glucocorticoid receptor; GRE, glucocorticoid responsive
elements; HSP90, heat-shock protein 90; Src, sarcoma kinase; MEK, mitogen-activated protein kinase kinase; PLCc, phosphoinositide-specic phospholipase c; JNK, c-Jun N-
terminal kinase; ERK, extracellular signal-regulated kinases; PKC, protein kinase C; PI3K, phosphoinositide 3-kinase; AKT, PTEN, phosphatase and tensin homolog.

cytoplasm or translocates to the nucleus (Rauch et al., 2010). In the that activated STAT5 and GR form complexes which bind DNA and
nucleus the GR acts as a transcriptional regulator of distinct GC- regulate gene expression independently of GREs. The idea that the
responsive target genes via direct DNA binding at glucocorticoid GR interacts with DNA-bound STAT5 as a cofactor was supported
response elements (GREs) as a dimer. Alternatively, GR can also by the nding that GR mutants decient for dimerization and
modulate the expression of genes through a GRE-independent DNA binding (GRdim) still synergize with STAT5 (Stoecklin et al.,
mechanism, which is mediated in part through proteinprotein 1997). More recently, it was shown that proteinprotein interac-
interactions with other transcription factors or coactivators tion of hepatic STAT5 and GR is essential for many of the functions
(Fig. 1) (Kassel and Herrlich, 2007; Beck et al., 2011). The degree exerted by either transcription factor in vivo (Engblom et al., 2007).
to which extent the GR dimerization or the monomeric activity Many functions of the GR depend on its cofactor interaction as re-
contributes to physiological effects of GCs varies dependent on vealed through the study of mice with a knock-in of the DBD-
the type of process studied (Rauch et al., 2010; Baschant et al., defective GRdim, which display relatively normal GH target gene
2011; Kleiman et al., 2011). expression pattern in liver (Reichardt et al., 1998; Tronche et al.,
2004). Whole genome expression analysis of STAT5, GR and
2.3. STAT5 and GR synergism in hepatocytes STAT5/GR knockout livers revealed that genes positively regulated
by either transcription factor overlap to a large extent (Engblom
DNA binding of the GR is believed to mediate most of its activat- et al., 2007). More than 40% of genes signicantly downregulated
ing function, while cross-talk with other transcription factors is in GR-decient livers were also downregulated in absence of
thought to mediate the repressive actions (Reichardt et al., 1998, STAT5. Correspondingly, almost 30% of genes downregulated in
2001). One of the exceptions, in which GR activates transcription STAT5-decient livers were also downregulated in the absence of
without classic DNA binding, is its functional interaction with GR. The magnitudes of expression changes highly correlated be-
STAT5. This synergism was rst shown for STAT5-dependent tran- tween all three genotypes and it became evident that STAT5GR
scription of the b-casein gene, which requires the GR as a transcrip- synergism preferentially affects gene sets involved in growth and
tional coactivator (Stoecklin et al., 1996, 1997). It became evident, maturation. Noteworthy, the expression changes elicited by STAT5
4 K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111

Fig. 2. STAT5 and GR proteinprotein interaction in postnatal body growth. (A) Structural properties of murine STAT5B and murine GR. The STAT5N-terminal domain
(tetramerization domain) and the AF1 domain of the GR are the protein-binding interfaces for the GR as a cofactor on DNA bound STAT5. (B) Schematic illustration of
postnatal body growth in GH transgenic mice, mice harboring a hepatic deletion of STAT5 in presence of the GH transgene, STAT5DN mice, GRdim mice and mice with a
hepatocyte-specic deletion of STAT5 or GR compared to wild type mice. STAT5, signal transducer and activator of transcription 5; GR, glucocorticoid receptor; SH2, Src
homology 2; AF-1, hormone-independent transactivation function domain; AF-2, transactivation domain; GH, growth hormone.

and the GR correlate closely with those found for various trunca- GH-signaling (Larons syndrome) (Laron et al., 1965; Rosenfeld
tions of the GHR (Rowland et al., 2005). Moreover, observed et al., 2007; Brooks and Waters, 2010), a major function of GH is
changes for male-predominant genes were in line with earlier re- regulation of longitudinal body growth as well as of all internal or-
ports (Clodfelter et al., 2006) which reported maturation-related gans excluding the brain. Here, mutations have been identied in
sexual dimorphic liver gene expression to be dependent on GH core genes of GH signaling, which affect the GHR itself, STAT5B,
action and hepatic STAT5. Further, whole genome expression anal- as well as IGF-1 (Rosenfeld et al., 2007; Brooks and Waters,
ysis of livers from mice expressing hypomorphic N-terminally trun- 2010). As veried by mouse knockout models, the regulation of
cated STAT5A and STAT5B proteins (referred to as STAT5DN mice) body size by GH is mainly executed by the activation of STAT5B
suggests that many of GH target genes depend on an intact which, in turn, mediates the transcription of Igf-1 and acid labile
STAT5N-terminus (Engblom et al., 2007). Indeed, in accordance subunit (Als) (Ooi et al., 1998; Woele et al., 2003a,b). IGF-1 and
with earlier studies STAT5GR synergism in hepatocytes was found ALS form a trimeric complex together with IGF-1 binding protein
to be independent of DNA-bound GR, but highly dependent on func- 3, termed bioactive IGF-1, in the serum (Dai and Baxter, 1994;
tional STAT5GR proteinprotein interaction (Figs. 1 and 2A). This Jones and Clemmons, 1995) to promote cellular growth and to con-
interaction requires the STAT5N-terminus and the AF-1 domain of trol neuroendocrine functions. The rst indication that STAT5B is
the GR as the protein-binding interfaces (Stoecklin et al., 1997; an essential mediator of postnatal body growth came from mice
Engblom et al., 2007). To date, there is little information whether which either lack the Stat5a or Stat5b gene. STAT5B-decient
other cell types or other cytokines and growth factors which signal males but not females suffer from impaired growth (Udy et al.,
through STAT5 display a similar STAT5GR cofactor interaction and 1997), while STAT5A-decient mice exhibit normal body stature
synergistic gene transcription apart from GH signaling. Future re- (Liu et al., 1997). The expression of N-terminally truncated STAT5A
search is required to elucidate if STAT5GR interaction is a selective and STAT5B proteins in STAT5DN mice affects postnatal body
requirement for the activation of specic gene sets in hepatocytes growth in either sex. However, STAT5DN females displayed more
or if that concept is of general importance for other cell types. pronounced dwarsm (Teglund et al., 1998). To evaluate the im-
pact of hepatic GH-STAT5 signaling and liver-derived IGF-1 on
3. STAT5 and GR function in body growth body growth, hepatocyte-specic knockout mice targeting the core
components of GHR-STAT5 signaling, the GHR (Fan et al., 2009),
As evidenced by patients with abnormally low circulating GH JAK2 (Sos et al., 2011) and STAT5 (Cui et al., 2007; Engblom
levels or mutations in core genes of the GH pathway disrupting et al., 2007; Friedbichler et al., 2012) were generated. All mice
K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111 5

decient in hepatic GHR-JAK2-STAT5 signaling are GH insensitive, In particular, we highlight those insights gained from hepatocyte-
with blunted hepatic Igf-1 mRNA expression and severely reduced specic deletions of both transcription factors in mice.
bioactive IGF-1 resulting in elevated plasma GH. Interestingly, de-
spite comparably reduced levels of circulating IGF-1, the pheno- 4.1. Glucose metabolism
types of the different mouse lines are quite diverse in regard to
postnatal body growth. Liver-specic deletion of STAT5 (Cui The liver is the main site for glucose storage in form of glycogen
et al., 2007) or the GHR (Fan et al., 2009) using a Cre recombinase and glucose synthesis via gluconeogenesis. The liver provides this
under the control of the albumin gene promoter (Alb-Cre (Yakar energy metabolite to maintain blood glucose level in times of need
et al., 1999)) did not lead to a reduction in body growth. Yet, and to provide glucose to extrahepatic tissues such as the brain,
deletion of STAT5 mediated by a hepatocyte-specic Cre recombi- which uses approximately 25% of total body glucose. Hepatic GC-
nase under the albumin gene promoter and the a-fetoprotein gene GR signaling plays a critical role in maintaining blood glucose level,
enhancer (Alfp-Cre (Kellendonk et al., 2000)) results in stunted particularly in states of energy deprivation, by directly controlling
body growth (Engblom et al., 2007), while Alb-Cre-mediated dele- rate limiting enzymes of gluconeogenesis such as PEPCK and
tion of JAK2 causes a modest but signicant decrease in body G6Pase (Hanson and Reshef, 1997; van Schaftingen and Gerin,
weight and length (Sos et al., 2011). The observed variations in 2002; Opherk et al., 2004). In turn, GC signaling impairs glucose
growth phenotypes remains poorly understood and may partly re- uptake in peripheral tissues, such as skeletal muscle and adipose
ect differences in gene deletion efciency and mouse genetic tissues. Hence, GCs oppose insulin action, which suppresses glu-
backgrounds. cose production by inhibiting hepatic glycogenolysis and gluco-
A recent study by our group using an Alfp-Cre-mediated STAT5 neogenesis, and stimulates glucose uptake, storage, and
knockout in the settings of systemic GH overexpression provided utilization by other tissues (Andrews and Walker, 1999). Thus, it
an additional hint that STAT5 signaling in liver is essential for is not surprising that pathologic conditions of elevated GC levels
GH-stimulated body growth (Friedbichler et al., 2012). Overexpres- are linked to defects in glucose metabolism characterized by insu-
sion of GH in mice leads to an alteration of body proportions lin resistance and hyperglycemia (Andrews and Walker, 1999;
resulting in an acromegaly-like phenotype and differential enlarge- Vegiopoulos and Herzig, 2007). In contrary, mice which are GR-
ment of internal organs (Eisen et al., 1998; Iida et al., 2004). In con- decient specically in liver show a mild decrease in blood glucose
trast, up to 9 weeks of age mice overexpressing GH but lacking level (Mueller et al., 2011) and suffer from profound hypoglycemia
hepatic STAT5 display body growth identical to that of wild type after prolonged fasting (Opherk et al., 2004). The inability to per-
(wt) littermates. Thereafter, STAT5 deciency in liver, despite the form de novo glucose synthesis is associated with a reduced induc-
presence of GH overexpression, results in growth retardation sim- tion of rate limiting enzymes of gluconeogenesis such as PEPCK
ilar to that caused by STAT5 deciency alone (Fig. 2). Noteworthy, (Opherk et al., 2004). Plasma GC levels were found to be markedly
despite the obvious reduction in body size, the bone length of ani- increased in response to fasting (Opherk et al., 2004), while we
mals systemically overexpressing GH yet lacking STAT5 in hepato- found an elevation of ACTH and GC levels in these mice already un-
cytes is 5% above that of wt littermates (Friedbichler et al., 2012). der basal conditions (Mueller et al., 2011). This elevation of sys-
These observations indicate that a direct action of GH on bone temic GCs in mutant animals might reect the bodys attempt to
and muscle and a paracrine function of IGF-1 (Yakar et al., 1999; sustain gluconeogenesis upon GR deciency. Interestingly, a simi-
Klover and Hennighausen, 2007) are not fully sufcient to compen- lar compensatory hypercortisolism is also reported in mice de-
sate for the loss of hepatic GH-STAT5 signaling. Of note, liver- cient for hexose-6-phosphate dehydrogenase. These mice lack
specic ablation of the GR leads to similar growth retardations as 11b-hydroxysteroid dehydrogenase type 1 reductase activity
observed for STAT5-decient mice while combined mutations do which leads to decreased intracellular levels of corticosterone
not add signicantly to the growth impairment caused by single and subsequent defects in glucose metabolism as well as impaired
mutations (Tronche et al., 2004; Engblom et al., 2007). Accordingly, responses of hepatic enzymes to fasting (Rogoff et al., 2007). A sec-
the growth retardation of STAT5DN mice is in line with a defect in ond possible cause for the compensatory activation of the HPA axis
GR cofactor recruitment, since the N-terminus of STAT5 needs to is an increased expression and release of liver-derived corticoste-
physically recruit the GR for proteinprotein interaction and gene roid binding globulin (CBG), which binds the majority of GCs in
regulation (Fig. 2) (Engblom et al., 2007). the circulation to retain them in a biologically inactive form
(Rosner, 1990). GCs suppress CBG expression, hence, GR knockout
mice show increased hepatic CBG expression (Mueller et al., 2011)
and display high basal CBG levels that are not suppressed by the
4. GH-STAT5 and GR function in metabolism and hepatic synthetic glucocorticoid Dexamethasone (Cole et al., 1999). The in-
steatosis crease in CBG and a decrease in free GCs might play a role in ele-
vated HPA activity due to decreased GC bioavailability, as
The liver plays a central role in the control of glucose and lipid observed in studies with gonadectomized rats and in a porcine
metabolism as it is the major site for interconversion, distribution model (Ousova et al., 2004; Viau and Meaney, 2004).
and storage of energy metabolites. Imbalances in hepatic metabolic Further, in response to hepatic GR deciency and the concomi-
function are tightly linked to non-alcoholic fatty liver disease tant impairment of the liver to counteract low energy levels by glu-
(NAFLD) and associated metabolic disorders (Feldstein, 2010; Bech- coneogenesis, a compensatory increase in glucagon (Opherk et al.,
mann et al., 2011). At this, GH and GC action is not only essential for 2004) and a parallel decrease in insulin were observed (Opherk
normal development and survival; it is also required for mainte- et al., 2004; Mueller et al., 2011). Elevated hepatic gluconeogenesis
nance of the bodys overall metabolic homeostasis (Vegiopoulos is a major contributor to hyperglycemia in type 2 diabetes. Here,
and Herzig, 2007; Moller and Jorgensen, 2009). Further, whole the states of insulin resistance or deciency favor glucose synthesis
genome expression analysis of gene networks affected by GH and by increased GC levels. Indeed, enhanced GR-dependent activation
GC signaling in liver has shown that STAT5 and GR control many as- of Pepck expression is reported in some murine models of obesity
pects of hepatocyte metabolism (Phuc Le et al., 2005; Cui et al., and diabetes, which can be limited by GR antagonism (Liu et al.,
2007; Engblom et al., 2007; Schirra et al., 2008). In the following 2006, 2008). In addition, different approaches using antisense oli-
paragraphs we discuss the current insights into the regulation of gonucleotide-mediated downregulation of GR expression show
glucose and lipid metabolism by GH-STAT5 and GC-GR signaling. that this improves fasting hyperglycemia and systemic glucose
6 K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111

homeostasis in diabetic mice without affecting blood GC levels 4.2. Lipid metabolism and hepatic steatosis
(Liang et al., 2005; Watts et al., 2005). Systemic GR antagonism
and the antisense oligonucleotide approach does not allow to Hepatic triglyceride (TG) stores are determined by the balance
determine inhibition of hepatic GC-GR signaling on diabetes of fatty acid (FA) uptake and release, de novo lipogenesis, and oxi-
related hyperglycemia, as GR signaling in other organs and con- dative clearance, a complex process regulated at pre-receptor,
comitant inhibition of glucose uptake, is most likely also impaired. transcriptional and posttranscriptional levels (Browning and Hor-
An indication for hepatic GR as a critical inducer of diabetic hyper- ton, 2004; Desvergne et al., 2006). Defects in GH and GC signaling
glycemia came from streptozotocin-induced diabetes in hepato- pathways have been implicated in NAFLD development. Chroni-
cyte-specic GR knockout mice, which display less severe cally increased GC levels are associated with a fatty liver pheno-
hyperglycemia and lack hepatic Pepck mRNA expression upon type in NAFLD (Targher et al., 2006). Moreover, fatty
diabetic challenge (Opherk et al., 2004). degeneration of hepatocytes was demonstrated in patients with
GH has both chronic and acute effects on glucose metabolism. Cushings syndrome (Shibli-Rahhal et al., 2006) and fatty liver dis-
The latter are designated as temporary insulin-like effects, and their ease is also a typical side effect of long-term systemic GC treat-
physiological signicance is not clear. The well studied chronic ment, e.g. immunosuppressive therapy (Schacke et al., 2002). GH
effects of GH oppose, like those of GCs, insulin action on glucose has pronounced lipolytic effects and under conditions of food
metabolism (Davidson, 1987). Excess GH is associated with im- deprivation provides peripheral tissues with ketone bodies as an
paired glucose tolerance, compensatory hyperinsulinemia, insulin alternative energy source to glucose. Excess GH, as observed in un-
resistance and fasting hyperglycemia. GH deciency, on the other treated acromegaly, is associated with increased lipolysis, de-
hand, is linked to enhanced insulin sensitivity, decreased fasting creased fat mass and an abnormal lipid prole (Moller and
glucose levels, decreased insulin secretion, and lowered hepatic Jorgensen, 2009). GHR loss of function mutations, on the other
glucose production (Gorin et al., 1990; Dominici and Turyn, 2002; hand, causes NAFLD in adults (Laron et al., 2008) and patients
Moller and Jorgensen, 2009). Upon systemic GHR deciency, as who are GH decient also display fatty degeneration of the liver
found in Ghr-null mice, GH is secreted in large quantities, but it more frequently than those with normal GH levels (Ichikawa
lacks biological effects due to deciency of its receptor (Zhou et al., 2003; Adams et al., 2004). Indeed, a recent case report has
et al., 1997). In contrast, Ames dwarf mice (Prop1df/df) suffer from shown that administration of recombinant GH can improve NAFLD
primary pituitary deciency and lack GH secretion from the anterior in a GH-decient patient accompanied by a reduction in brosis
pituitary (Sornson et al., 1996). However, both Ghr-null and Ames and reversion of hepatocyte ballooning (Takahashi et al., 2007).
dwarf mice exhibit a state of hypersensitivity to insulin and an in- Even though, aberrant GC and GH action is frequently associ-
creased hypoglycemic response to exogenous insulin (Dominici ated with steatosis and associated metabolic disorders, the impact
et al., 2002; Dominici and Turyn, 2002). The counterpart to the stud- of GH-STAT5 and GC-GR signaling on liver lipid metabolism, partic-
ies described above is the bovine GH transgenic mouse model, ularly in humans, is less well characterized. However, several re-
which displays hyperinsulinemia and insulin resistance in response cent studies in mice have provided new mechanistic insights.
to excess GH exposure, but nearly normal glucose tolerance (Valera Adult mice with a hepatocyte-specic GR knockout display an in-
et al., 1993; Balbis et al., 1996). Additionally, hepatocyte-specic crease in body fat, while plasma TG levels are substantially lower
impairment of GH-STAT5 signaling by targeting the GHR or STAT5 (Opherk et al., 2004). The relative size of GR-decient livers as well
induces a state of insulin resistance, which manifests itself in pro- as liver histology is comparable to those of wt mice (Mueller et al.,
found hyperinsulinemia and glucose intolerance (Cui et al., 2007; 2011). Yet, an additional study of these mice has shown that GR
Fan et al., 2009; Mueller et al., 2011). In regard to the induction of signaling promotes transient hepatic fat accumulation following
an insulin resistant state, interference with hepatic GH-STAT5 sig- partial hepatectomy (Shteyer et al., 2004). Interestingly, Herzig
naling closely resembles the anti-insulin actions described in GH and colleagues (Lemke et al., 2008) have shown that liver specic
transgenic mice. Here, chronic GH excess results in a diminished re- knockdown of the GR, mediated by adenoviral shRNA, improves
sponse to insulin injection particularly in skeletal muscle at the le- the steatotic phenotype in mouse models of fatty liver disease.
vel of IRS-1 phosphorylation and downstream signaling events such Here, hepatic GR signaling upon metabolic challenge was linked
as PI3K activation (Dominici and Turyn, 2002). More recently, ex- to downregulation of genes involved in hepatic TG lipolysis and
cess GH exposure in mice and the acquired insulin resistance was b-oxidation, which is accompanied by upregulation of fatty acid
linked to enhanced expression of p85a regulatory subunit of PI3K uptake and storage. Moreover, the GR-dependent hepatic lipid
accompanied by a decrease in IRS-1-associated PI3K activity in the accumulation was shown to depend on inhibition of expression
skeletal muscle and white adipose tissue (Barbour et al., 2005; del of Hes-1, a known anti-lipogenic factor (Herzig et al., 2003). Resto-
Rincon et al., 2007). Hepatocyte-specic impairment of GH-STAT5 ration of Hes-1 expression favorable changes hepatic lipid metab-
signaling induces GH insensitivity of the liver and a corresponding olism including the downregulation of Pparc, Cd36 and caveolin
elevation of pituitary GH secretion. As GH signaling in other tissues expression, which results in lowered hepatic TG content (Lemke
remains intact, it is tempting to speculate that a defective insulin et al., 2008).
receptor signaling in muscle and adipose tissue as observed in GH In contrast, mouse models of GH excess are not steatotic and
overexpressing mice partly accounts for insulin resistance. More- they have lower hepatic TG content (Wang et al., 2007; Friedbichler
over, deletion of hepatic STAT5 markedly impairs downstream insu- et al., 2012), whereas impaired GH-STAT5 signaling in liver perturbs
lin signaling in the liver (Mueller et al., 2011) and favors an lipid metabolism resulting in liver steatosis even upon excess GH
upregulation of hepatic p85a mRNA expression (Mueller et al., exposure (Friedbichler et al., 2012). Several studies have shed light
unpublished observation). A predominant role for defective hepatic on the underlying mechanisms how hepatic GHRSTAT5 signaling
insulin signaling is also described in mice with liver-specic knock- maintains hepatic lipid homeostasis. Hepatocyte-specic ablation
out of IR, where impaired IR signal transduction contributes to fast- of the GHR (Fan et al., 2009), JAK2 (Sos et al., 2011) and STAT5
ing hyperglycemia (Michael et al., 2000). Interestingly, in the (Cui et al., 2007; Mueller et al., 2011; Friedbichler et al., 2012) re-
settings of hepatic STAT5 deciency and the associated GH insensi- sults in progressive steatosis accompanied by elevated liver dam-
tivity of the liver, the additional lack of GR in hepatocytes is not age parameters. The increase in TG accumulation in the absence
sufcient to ameliorate hyperglycemia. In summary, STAT5 and of hepatic GH-STAT5 signaling probably results from an upregula-
STAT5/GR knockout mice are equally affected by hyperinsulinemia tion of genes involved in hepatic fatty acid uptake and/or de novo
and insulin resistance (Mueller et al., 2011). synthesis. Genetic alterations shared by the before mentioned
K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111 7

Fig. 3. Schematic illustrations demonstrating the development of liver phenotypes following combined hepatic STAT5/GR deletion and STAT5 deletion in GH transgenic mice.
Loss of hepatic GH-STAT5 signaling, in presence and absence of hepatic GR, causes defects in lipid and glucose metabolism leading to steatosis, hyperglycemia, insulin
resistance and impaired hepatic insulin receptor signal transduction. On the molecular level, impairment of hepatic GH-STAT5 signaling is associated with enhanced pro-
lipogenic PPARc and SREBP-1c signaling leading to increased lipogenesis and/or increased hepatic FFA uptake (1; see text for details). Further, loss of STAT5 results in hepatic
GH insensitivity resulting in high endogenous GH levels, while hepatic GR deciency leads to hypercortisolism. Consequently, high GH levels (endogenous and transgene
expression; 1) or high endogenous GH level in combination with elevated GCs (1 and 2) lead to lipolysis of adipose tissues and enhanced FFA release. The increase in
circulating FFA in combination with enhanced expression of the fatty acid transporter CD36 results in additional FA accumulation in the liver. The increased hepatic FA load
and associated metabolic dysfunctions upon STAT5/GR deciency as well as STAT5 deciency in GH transgenic mice results in oxidative stress and chronic liver damage.
These metabolic changes additionally activate the stress kinase signaling (JNK1 and/or p38). Moreover, increased lipid synthesis, deregulated expression of hepatoprotective
factors (EGFR, PRLR, LIFR; HNF6), accumulation of mutations (increased DNA damage) following loss of cell cycle control (reduced p53 levels), and enhanced activity of
tumor-promoting STAT3 and c-JUN (elevated protein levels induced by STAT3 and increased phosphorylation mediated by JNK1 and p38 stress kinases) within hepatocytes
contribute to severe tissue damage and the development of HCC. STAT, signal transducer and activator of transcription; GR, glucocorticoid receptor; GH, growth hormone;
GCs, glucocorticoids; GHR, GH receptor; PPARc, peroxisome proliferator-activated receptor gamma; SREBP-1c, sterol regulatory element-binding protein 1c; FFA, free fatty
acids; CD36, cluster of differentiation 36; JNK1, c-JUN-N-terminal kinase; EGFR, epithelial growth factor receptor; PRLR, prolactin receptor; LIFR, leukemia inhibitory factor
receptor; HNF6, hepatocyte nuclear factor 6; ROS, reactive oxygen species.

models include enhanced expression of Pparc (FA uptake and syn- Vice versa, overexpression of bovine GH reduces expression of
thesis) and its target gene Cd36 (FA uptake), which are both fre- Srebp-1c and several lipogenic downstream target genes in liver, de-
quently associated with development of fatty liver disorders spite overt hyperinsulinemia, a well known cause of increased
(Fig. 3) (Browning and Horton, 2004; Bechmann et al., 2011). It Srebp-1c transcription (Olsson et al., 2003). Consistently, GH treat-
was shown that treatment with a PPARc-specic antagonist leads ment of wt mice leads to decreased hepatic Srebp-1c expression,
to reduced Cd36 expression and decreased lipid load in JAK2-de- and GH-activated STAT5 was found to interact with the Srebp-1c
cient livers (Sos et al., 2011). A further study has established that gene promoter. Thereby, it might contribute to a transcriptional
STAT5 binds to the Cd36 gene promoter which might suppress tran- inhibition (Mueller et al., 2011). Further, immature SREBP-1c can
scription (Barclay et al., 2011). Yet, upregulation of Pparc itself is be activated in response to decreased expression of Fgf21 and Insig2
presumably not due to loss of a STAT5-mediated inhibition. It is (Osborne and Espenshade, 2009; Xu et al., 2009), the transcription
rather a consequence of enhanced GH-dependent STAT1 activation of which is severely decreased in liver upon hepatocyte-specic
upon hepatic STAT5 deciency (Cui et al., 2007; Barclay et al., 2011). STAT5 deletion and upon impairment of GHR signaling (Barclay
Secondly, enhanced expression of the prolipogenic transcription et al., 2011; Mueller et al., 2011). Overall, the precise mechanisms
factor SREBP-1c (Browning and Horton, 2004; Bechmann et al., leading to deregulation of PPARc and SREBP-1c signaling upon im-
2011) is a possible cause for steatosis in the absence of GH-STAT5 paired hepatic GH-STAT5 signaling are not completely understood.
signaling (Fig. 3). Hepatic GHR and STAT5 deciency was shown Interestingly, the additional deletion of hepatic GR neither ame-
to result in enhanced expression of Srebp-1c and lipogenic down- liorates the steatosis phenotype caused by STAT5 deciency nor
stream targets such as Fas (Fan et al., 2009; Mueller et al., 2011). does it improves the gene expression prole of altered hepatic lipid
8 K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111

metabolism, e.g. by downregulation of Pparc and Cd36 transcrip- FFA, TNF-a, and ROS levels are typically observed in NAFLD and
tion. A combined STAT5/GR deciency rather results in an even are associated with persistent hepatocyte damage (Bechmann
more pronounced hepatocyte TG accumulation (Mueller et al., et al., 2011). These conditions support the accumulation of DNA
2011). The increased hepatic TG load in hepatic STAT5/GR- damage and sustained stress-dependent JNK1 activity (Mueller
decient animals compared to STAT5-decient animals results et al., 2011), which are both recognized as critical factors in the
from a combination of elevated plasma GH and GC levels, which, promotion and progression of human and murine HCC (Luedde
in turn, activate STAT5 and GR signaling in adipocytes. Both tran- et al., 2007; Park et al., 2010; Starley et al., 2010).
scription factors drive the upregulation of adipose tissue triglycer- GH overexpression in mice is linked to a severe systemic
ide lipase and hormone sensitive lipase, which are essential for inammatory phenotype, reduced life expectancy and the develop-
lipolysis (Fain et al., 2008; Mueller et al., 2011). The subsequent in- ment of liver tumors (Orian et al., 1990; Bartke et al., 2002).
crease in available plasma FFA in combination with enhanced Intriguingly, hepatic STAT5 deciency reverses all pathologic alter-
expression of the fatty acid transporter CD36 in hepatocytes facil- ations induced by high GH levels, but leads to a more aggressive
itates additional TG accumulation in the liver (Fig. 3). form of HCC at earlier time points (Friedbichler et al., 2012). As
In conclusion, hepatic GH-STAT5 signaling protects the liver in the former model, hepatic STAT5 deciency attributes to in-
from steatosis by ensuring proper transcriptional control of genes creased adipose tissue derived FA accumulation in hepatocytes,
involved in regulation of hepatic de novo lipogenesis and FA subsequent chronic liver damage and accumulation of DNA dam-
uptake. age. Accordingly, JNK1 activity and a related increase in c-JUN
expression and activity were also present in this transgenic HCC
model. Further, following c-JUN activation (Eferl et al., 2003) or
5. GH-STAT5 signaling and HCC other oncogenic mechanisms potentially involving STAT3 (Niu
et al., 2005) p53 activity was abolished in STAT5-decient livers.
Hepatocellular carcinoma (HCC) is a common complication of Thereby, diminished activity of p53 downstream signaling might
chronic liver disease and in most cases carcinogenesis follows a impair clearance of cells harboring DNA damage and favor subse-
sequential process, with cirrhosis as an intermediate key step quent xation of mutations. In this regard, it was shown that per-
(Feldstein, 2010; Bechmann et al., 2011). However, development sistent activation of STAT5A triggers a permanent cell cycle arrest
of HCC is increasingly observed in absence of advanced liver injury with characteristics of cellular senescence including activation of
and cirrhosis (Paradis et al., 2009; Starley et al., 2010; Bechmann p53 and a constitutive activation of the DNA damage response in
et al., 2011). In this regard, independent risk factors for HCC in- non-hepatic cells (Mallette et al., 2007a,b; Calabrese et al., 2009).
clude obesity and diabetes (Calle et al., 2003; Paradis et al., 2009). Based on the current state of knowledge, HCC development in
STAT5 activation is oncogenic in hematopoietic cancers and it the absence of STAT5 might be the result of several direct and indi-
correlates with poor prognosis in myeloid leukemia. However, rect mechanisms: (1) Deregulation of STAT5 target genes involved
the role of STAT5 protein activation in carcinomas is more compli- in the protection of hepatocyte integrity and cell cycle inhibition.
cated and cell type specic. Persistent STAT5 activation correlates (2) Increased STAT3 signaling most likely due to misrecruitment
with a good prognosis in breast cancer, while in prostate cancer to the GHR in absence of STAT5 accelerates progression of chronic
the opposite was reported (reviewed in Ferbeyre and Moriggl liver disease. (3) Metabolic dysfunctions contribute to liver cell and
(2011)). So far, there is little information regarding the impact of DNA damage and subsequent activation of stress-kinase signaling.
hepatic GH-STAT5 signaling on HCC development. Increased (4) An accumulation of mutations is most likely facilitated by a loss
STAT5B activity was reported to correlate with more aggressive tu- of cell-cycle control and tumor-suppressive functions of p53
mors and poor clinical outcomes in hepatitis B virus-related HCC (Fig. 3). In summary, while some mechanisms by which GH-
due to increased cell motility and concomitant tumor spread (Lee activated STAT5 contributes to safeguard mechanisms that protect
et al., 2006). In patients with liver cirrhosis, a premalignant condi- hepatocytes from chronic injury and tumorigenic transformation
tion, the GH-IGF-1 axis is known to be severely impaired caused by have been revealed, further work is clearly required to better
a state of acquired hepatic GH resistance (Picardi et al., 2006), understand the consequences and possible connection to develop-
which is indicative of low STAT5 activity. In parallel, a mouse mod- ment of human disease.
el of cholestatic liver disease with ablated hepatic GH-STAT5
signaling displayed an early and more severe liver brosis pheno-
type. This was attributed to the lack of IGF-1 and down-regulation 6. Concluding remarks
of hepatoprotective factors such as Egfr, Lifr, Plr and Hnf-6 (Fig. 3)
(Blaas et al., 2010). Upon CCl4 challenge, hepatic STAT5 deciency Conditional knockouts targeting the core components of murine
promotes the development of liver brosis and, in some cases, HCC hepatic GH-STAT5 signaling and the GR have provided an impor-
as a result of increased STAT3 activation and TGF-b stabilization tant tool to obtain mechanistic insights into their role in liver func-
(Hosui et al., 2009). STAT5 activity in hepatocytes was suggested tion. We provided an overview of overlapping and distinct
to promote cell cycle arrest upon chronic hepatocyte injury, while functions of hepatic GH-STAT5 and GC-GR signaling in postnatal
loss of STAT5 signaling favors activation of pro-survival and prolif- body growth, glucose/lipid homeostasis and metabolic diseases.
eration pathways. This was linked to (1) reduced expression of the Aberrant hepatic lipid and glucose metabolism are closely re-
cell cycle inhibitors and STAT5 target genes Cdkn1a and Cdkn2b, lated to the pathogenesis of the most common liver diseases
and (2) excessive GH-dependent activation of STAT3 in absence including their progression to hepatocarcinogenesis. Better under-
of STAT5 (Hosui et al., 2009; Yoo et al., 2011; Yu et al., 2011). Upon standing of the underlying molecular mechanisms is thus crucial,
additional challenge, in form of increased adipose tissue derived particularly in light of the increasing prevalence of obesity and
FA inux due to combined GH resistance and hypercortisolism its pathological consequences.
(Mueller et al., 2011) or GH overexpression (Friedbichler et al., A characteristic of metabolic liver disease in the absence of he-
2012), STAT5-decient livers develop HCC in the presence of pro- patic GH-STAT5/GR signaling is the activation of both signaling
gressive steatosis, despite minor inammation and brotic degen- pathways in peripheral tissues such as the adipose compartments.
eration. Steatosis combined with elevated plasma FFAs was shown Thus, the use of transgenic mice will greatly help to understand the
to coincide with increased plasma and liver TNF-a level and oxida- contribution of STAT5 and GR signaling in extra-hepatic tissues to
tive stress in hepatocytes (Mueller et al., 2011). An elevation of the aberrations in carbohydrate and lipid metabolism. Addition-
K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111 9

ally, the downstream molecular mechanisms of GH-STAT5 and GC- dependent activation of male genes and repression of female genes revealed
by microarray analysis. Mol. Endocrinol. 20, 13331351.
GR signaling particularly in hepatic lipid metabolism and steatosis
Cole, T.J., Blendy, J.A., Monaghan, A.P., Krieglstein, K., Schmid, W., Aguzzi, A.,
needs to be further analyzed. While interference with hepatic GR Fantuzzi, G., Hummler, E., Unsicker, K., Schutz, G., 1995. Targeted disruption of
signaling seems to ameliorate TG accumulation in mouse models the glucocorticoid receptor gene blocks adrenergic chromafn cell development
of fatty liver disease, a protective role for GH-STAT5 signaling and severely retards lung maturation. Genes Develop. 9, 16081621.
Cole, T.J., Harris, H.J., Hoong, I., Solomon, N., Smith, R., Krozowski, Z., Fullerton, M.J.,
has emerged in chronic liver disease including HCC development. 1999. The glucocorticoid receptor is essential for maintaining basal and
HCC is a complex, heterogeneous cancer, which develops in a mul- dexamethasone-induced repression of the murine corticosteroid-binding
tistep process involving deregulation of multiple cellular signaling globulin gene. Mol. Cell. Endocrinol. 154, 2936.
Cui, Y., Riedlinger, G., Miyoshi, K., Tang, W., Li, C., Deng, C.X., Robinson, G.W.,
pathways and impaired tissue homeostasis. Hence, future studies Hennighausen, L., 2004. Inactivation of Stat5 in mouse mammary epithelium
on global gene expression proling of HCCs will help to pursue during pregnancy reveals distinct functions in cell proliferation, survival, and
the synergism of impaired GH-STAT5 signaling and potentially differentiation. Mol. Cell. Biol. 24, 80378047.
Cui, Y., Hosui, A., Sun, R., Shen, K., Gavrilova, O., Chen, W., Cam, M.C., Gao, B.,
deregulated tumor suppressor as well as oncogenic pathways in Robinson, G.W., Hennighausen, L., 2007. Loss of signal transducer and activator
hepatocarcinogenesis. of transcription 5 leads to hepatosteatosis and impaired liver regeneration.
Hepatology 46, 504513.
Dai, J., Baxter, R.C., 1994. Regulation in vivo of the acid-labile subunit of the rat
Acknowledgments serum insulin-like growth factor-binding protein complex. Endocrinology 135,
23352341.
Davidson, M.B., 1987. Effect of growth hormone on carbohydrate and lipid
Recent work by the authors was supported by Grant SFB F28
metabolism. Endocr. Rev. 8, 115131.
from the Austrian Science Funds (FWF; Project F2807-B20) to del Rincon, J.P., Iida, K., Gaylinn, B.D., McCurdy, C.E., Leitner, J.W., Barbour, L.A.,
R.M., K.M., M.T., K.F. and J.W.K. H.E. was supported by grants of Kopchick, J.J., Friedman, J.E., Draznin, B., Thorner, M.O., 2007. Growth hormone
the Vienna Science and Technology Fund (WWTF Project LS07- regulation of p85alpha expression and phosphoinositide 3-kinase activity in
adipose tissue: mechanism for growth hormone-mediated insulin resistance.
058). J.P.T. was supported by the Boehringer Stiftung, the Deutsche Diabetes 56, 16381646.
Forschungsgemeinschaft (TU220/3 and TU220/6 and by the Euro- Desvergne, B., Michalik, L., Wahli, W., 2006. Transcriptional regulation of
pean Union FP7-BrainAge). metabolism. Physiol. Rev. 86, 465514.
Dominici, F.P., Turyn, D., 2002. Growth hormone-induced alterations in the insulin-
signaling system. Exp. Biol. Med. (Maywood) 227, 149157.
References Dominici, F.P., Hauck, S., Argentino, D.P., Bartke, A., Turyn, D., 2002. Increased
insulin sensitivity and upregulation of insulin receptor, insulin receptor
substrate (IRS)-1 and IRS-2 in liver of Ames dwarf mice. J. Endocrinol. 173,
Adams, L.A., Feldstein, A., Lindor, K.D., Angulo, P., 2004. Nonalcoholic fatty liver
8194.
disease among patients with hypothalamic and pituitary dysfunction.
Eferl, R., Ricci, R., Kenner, L., Zenz, R., David, J.P., Rath, M., Wagner, E.F., 2003. Liver
Hepatology 39, 909914.
tumor development: c-Jun antagonizes the proapoptotic activity of p53. Cell
Andrews, R.C., Walker, B.R., 1999. Glucocorticoids and insulin resistance: old
112, 181192.
hormones, new targets. Clin. Sci. (Lond.) 96, 513523.
Eisen, E.J., Peterson, C.B., Parker, I.J., Murray, J.D., 1998. Effects of zinc ion
Balbis, A., Bartke, A., Turyn, D., 1996. Overexpression of bovine growth hormone in
concentration on growth, fat content and reproduction in oMT1a-oGH
transgenic mice is associated with changes in hepatic insulin receptors and in
transgenic mice. Growth Devlop. Aging 62, 173186.
their kinase activity. Life Sci. 59, 13631371.
Engblom, D., Kornfeld, J.W., Schwake, L., Tronche, F., Reimann, A., Beug, H.,
Barbour, L.A., Mizanoor Rahman, S., Gurevich, I., Leitner, J.W., Fischer, S.J., Roper,
Hennighausen, L., Moriggl, R., Schutz, G., 2007. Direct glucocorticoid receptor-
M.D., Knotts, T.A., Vo, Y., McCurdy, C.E., Yakar, S., Leroith, D., Kahn, C.R., Cantley,
Stat5 interaction in hepatocytes controls body size and maturation-related gene
L.C., Friedman, J.E., Draznin, B., 2005. Increased P85alpha is a potent negative
expression. Genes Devlop. 21, 11571162.
regulator of skeletal muscle insulin signaling and induces in vivo insulin
Fain, J.N., Cheema, P., Tichansky, D.S., Madan, A.K., 2008. Stimulation of human
resistance associated with growth hormone excess. J. Biol. Chem. 280, 37489
omental adipose tissue lipolysis by growth hormone plus dexamethasone. Mol.
37494.
Cell. Endocrinol. 295, 101105.
Barclay, J.L., Nelson, C.N., Ishikawa, M., Murray, L.A., Kerr, L.M., McPhee, T.R., Powell,
Fan, Y., Menon, R.K., Cohen, P., Hwang, D., Clemens, T., Digirolamo, D.J., Kopchick, J.J.,
E.E., Waters, M.J., 2011. GH-dependent STAT5 signaling plays an important role
Leroith, D., Trucco, M., Sperling, M.A., 2009. Liver-specic deletion of the growth
in hepatic lipid metabolism. Endocrinology 152, 181192.
hormone receptor reveals essential role of GH signaling in hepatic lipid
Bartke, A., Chandrashekar, V., Bailey, B., Zaczek, D., Turyn, D., 2002. Consequences of
metabolism. J. Biol. Chem.
growth hormone (GH) overexpression and GH resistance. Neuropeptides 36,
Feldstein, A.E., 2010. Novel insights into the pathophysiology of nonalcoholic fatty
201208.
liver disease. Semin. Liver Dis. 30, 391401.
Baschant, U., Frappart, L., Rauchhaus, U., Bruns, L., Reichardt, H.M., Kamradt, T.,
Ferbeyre, G., Moriggl, R., 2011. The role of Stat5 transcription factors as tumor
Brauer, R., Tuckermann, J.P., 2011. Glucocorticoid therapy of antigen-induced
suppressors or oncogenes. Biochim. Biophys. Acta 1815, 104114.
arthritis depends on the dimerized glucocorticoid receptor in T cells. Proc. Natl.
Flores-Morales, A., Greenhalgh, C.J., Norstedt, G., Rico-Bautista, E., 2006. Negative
Acad. Sci. USA 108, 1931719322.
regulation of growth hormone receptor signaling. Mol. Endocrinol. 20, 241253.
Bechmann, L.P., Hannivoort, R.A., Gerken, G., Hotamisligil, G.S., Trauner, M., Canbay,
Friedbichler, K., Themanns, M., Mueller, K.M., Schlederer, M., Kornfeld, J.W.,
A., 2011. The interaction of hepatic lipid and glucose metabolism in liver
Terracciano, L.M., Kozlov, A.V., Haindl, S., Kenner, L., Kolbe, T., Mueller, M.,
diseases. J. Hepatol.
Snibson, K.J., Heim, M.H., Moriggl, R., 2012. Growth hormone-induced STAT5
Beck, I.M., De Bosscher, K., Haegeman, G., 2011. Glucocorticoid receptor mutants:
signaling causes gigantism, inammation and premature death but protects
man-made tools for functional research. Trends Endocrinol. Metab. 22, 295
mice from aggressive liver cancer. Hepatology.
310.
Gorin, E., Tai, L.R., Honeyman, T.W., Goodman, H.M., 1990. Evidence for a role of
Blaas, L., Kornfeld, J.W., Schramek, D., Musteanu, M., Zollner, G., Gumhold, J., van Zijl,
protein kinase C in the stimulation of lipolysis by growth hormone and
F., Schneller, D., Esterbauer, H., Egger, G., Mair, M., Kenner, L., Mikulits, W., Eferl,
isoproterenol. Endocrinology 126, 29732982.
R., Moriggl, R., Penninger, J., Trauner, M., Casanova, E., 2010. Disruption of the
Hanson, R.W., Reshef, L., 1997. Regulation of phosphoenolpyruvate carboxykinase
growth hormonesignal transducer and activator of transcription 5insulinlike
(GTP) gene expression. Annu. Rev. Biochem. 66, 581611.
growth factor 1 axis severely aggravates liver brosis in a mouse model of
Hennighausen, L., Robinson, G.W., 2008. Interpretation of cytokine signaling
cholestasis. Hepatology 51, 13191326.
through the transcription factors STAT5A and STAT5B. Genes Devlop. 22,
Brooks, A.J., Waters, M.J., 2010. The growth hormone receptor: mechanism of
711721.
activation and clinical implications. Nat. Rev. Endocrinol. 6, 515525.
Herzig, S., Hedrick, S., Morantte, I., Koo, S.H., Galimi, F., Montminy, M., 2003. CREB
Browning, J.D., Horton, J.D., 2004. Molecular mediators of hepatic steatosis and liver
controls hepatic lipid metabolism through nuclear hormone receptor PPAR-
injury. J. Clin. Invest. 114, 147152.
gamma. Nature 426, 190193.
Buckley, T.M., Schatzberg, A.F., 2005. On the interactions of the hypothalamic
Hosui, A., Kimura, A., Yamaji, D., Zhu, B.M., Na, R., Hennighausen, L., 2009. Loss of
pituitaryadrenal (HPA) axis and sleep: normal HPA axis activity and circadian
STAT5 causes liver brosis and cancer development through increased TGF-
rhythm, exemplary sleep disorders. J. Clin. Endocrinol. Metab. 90, 31063114.
{beta} and STAT3 activation. J. Exp. Med.
Calabrese, V., Mallette, F.A., Deschenes-Simard, X., Ramanathan, S., Gagnon, J.,
Ichikawa, T., Hamasaki, K., Ishikawa, H., Ejima, E., Eguchi, K., Nakao, K., 2003. Non-
Moores, A., Ilangumaran, S., Ferbeyre, G., 2009. SOCS1 links cytokine signaling
alcoholic steatohepatitis and hepatic steatosis in patients with adult onset
to p53 and senescence. Mol. Cell. 36, 754767.
growth hormone deciency. Gut 52, 914.
Calle, E.E., Rodriguez, C., Walker-Thurmond, K., Thun, M.J., 2003. Overweight,
Iida, K., Del Rincon, J.P., Kim, D.S., Itoh, E., Nass, R., Coschigano, K.T., Kopchick, J.J.,
obesity, and mortality from cancer in a prospectively studied cohort of US
Thorner, M.O., 2004. Tissue-specic regulation of growth hormone (GH)
adults. New Engl. J. Med. 348, 16251638.
receptor and insulin-like growth factor-I gene expression in the pituitary and
Clodfelter, K.H., Holloway, M.G., Hodor, P., Park, S.H., Ray, W.J., Waxman, D.J., 2006.
liver of GH-decient (lit/lit) mice and transgenic mice that overexpress bovine
Sex-dependent liver gene expression is extensive and largely dependent upon
GH (bGH) or a bGH antagonist. Endocrinology 145, 15641570.
signal transducer and activator of transcription 5b (STAT5b): STAT5b-
10 K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111

Jeay, S., Sonenshein, G.E., Postel-Vinay, M.C., Kelly, P.A., Baixeras, E., 2002. Growth Niu, G., Wright, K.L., Ma, Y., Wright, G.M., Huang, M., Irby, R., Briggs, J., Karras, J.,
hormone can act as a cytokine controlling survival and proliferation of immune Cress, W.D., Pardoll, D., Jove, R., Chen, J., Yu, H., 2005. Role of Stat3 in regulating
cells: new insights into signaling pathways. Mol. Cell. Endocrinol. 188, 17. p53 expression and function. Mol. Cell. Biol. 25, 74327440.
Jones, J.I., Clemmons, D.R., 1995. Insulin-like growth factors and their binding Olsson, B., Bohlooly, Y.M., Brusehed, O., Isaksson, O.G., Ahren, B., Olofsson, S.O.,
proteins: biological actions. Endocr. Rev. 16, 334. Oscarsson, J., Tornell, J., 2003. Bovine growth hormone-transgenic mice have
Kassel, O., Herrlich, P., 2007. Crosstalk between the glucocorticoid receptor and major alterations in hepatic expression of metabolic genes. Am. J. Physiol.
other transcription factors: molecular aspects. Mol. Cell. Endocrinol. 275, 13 Endocrinol. Metab. 285, E504-11.
29. Ooi, G.T., Hurst, K.R., Poy, M.N., Rechler, M.M., Boisclair, Y.R., 1998. Binding of
Kellendonk, C., Opherk, C., Anlag, K., Schutz, G., Tronche, F., 2000. Hepatocyte- STAT5a and STAT5b to a single element resembling a gamma-interferon-
specic expression of Cre recombinase. Genesis 26, 151153. activated sequence mediates the growth hormone induction of the mouse acid-
Kleiman, A., Hubner, S., Rodriguez Parkitna, J.M., Neumann, A., Hofer, S., Weigand, labile subunit promoter in liver cells. Mol. Endocrinol. 12, 675687.
M.A., Bauer, M., Schmid, W., Schutz, G., Libert, C., Reichardt, H.M., Tuckermann, Opherk, C., Tronche, F., Kellendonk, C., Kohlmuller, D., Schulze, A., Schmid, W.,
J.P., 2011. Glucocorticoid receptor dimerization is required for survival in septic Schutz, G., 2004. Inactivation of the glucocorticoid receptor in hepatocytes leads
shock via suppression of interleukin-1 in macrophages. FASEB J. 26, 722729. to fasting hypoglycemia and ameliorates hyperglycemia in streptozotocin-
Klover, P., Hennighausen, L., 2007. Postnatal body growth is dependent on the induced diabetes mellitus. Mol. Endocrinol. 18, 13461353.
transcription factors signal transducers and activators of transcription 5a/b in Orian, J.M., Tamakoshi, K., Mackay, I.R., Brandon, M.R., 1990. New murine model for
muscle: a role for autocrine/paracrine insulin-like growth factor I. hepatocellular carcinoma: transgenic mice expressing metallothionein-ovine
Endocrinology 148, 14891497. growth hormone fusion gene. J. Natl. Cancer Inst. 82, 393398.
Kornfeld, J.W., Grebien, F., Kerenyi, M.A., Friedbichler, K., Kovacic, B., Zankl, B., Osborne, T.F., Espenshade, P.J., 2009. Evolutionary conservation and adaptation in
Hoelbl, A., Nivarti, H., Beug, H., Sexl, V., Muller, M., Kenner, L., Mullner, E.W., the mechanism that regulates SREBP action: what a long, strange tRIP its been.
Gouilleux, F., Moriggl, R., 2008. The different functions of Stat5 and chromatin Genes Devlop. 23, 25782591.
alteration through Stat5 proteins. Front. Biosci. 13, 62376254. Ousova, O., Guyonnet-Duperat, V., Iannuccelli, N., Bidanel, J.P., Milan, D., Genet, C.,
Lamia, K.A., Papp, S.J., Yu, R.T., Barish, G.D., Uhlenhaut, N.H., Jonker, J.W., Downes, Llamas, B., Yerle, M., Gellin, J., Chardon, P., Emptoz-Bonneton, A., Pugeat, M.,
M., Evans, R.M., 2011. Cryptochromes mediate rhythmic repression of the Mormede, P., Moisan, M.P., 2004. Corticosteroid binding globulin: a new target
glucocorticoid receptor. Nature 480, 552556. for cortisol-driven obesity. Mol. Endocrinol. 18, 16871696.
Lanning, N.J., Carter-Su, C., 2006. Recent advances in growth hormone signaling. Paradis, V., Zalinski, S., Chelbi, E., Guedj, N., Degos, F., Vilgrain, V., Bedossa, P.,
Rev. Endocr. Metab. Disorders 7, 225235. Belghiti, J., 2009. Hepatocellular carcinomas in patients with metabolic
Laron, Z., Mannheimer, S., Guttmann, S., 1965. Plasma disappearance of various syndrome often develop without signicant liver brosis: a pathological
131-I-labelled growth hormone preparations in man and rabbit. Nature 207, analysis. Hepatology 49, 851859.
298299. Park, E.J., Lee, J.H., Yu, G.Y., He, G., Ali, S.R., Holzer, R.G., Osterreicher, C.H., Takahashi,
Laron, Z., Ginsberg, S., Webb, M., 2008. Nonalcoholic fatty liver in patients with H., Karin, M., 2010. Dietary and genetic obesity promote liver inammation and
Laron syndrome and GH gene deletion preliminary report. Growth Horm. IGF tumorigenesis by enhancing IL-6 and TNF expression. Cell 140, 197208.
Res. 18, 434438. Phuc Le, P., Friedman, J.R., Schug, J., Brestelli, J.E., Parker, J.B., Bochkis, I.M., Kaestner,
Laz, E.V., Sugathan, A., Waxman, D.J., 2009. Dynamic in vivo binding of STAT5 to K.H., 2005. Glucocorticoid receptor-dependent gene regulatory networks. PLoS
growth hormone-regulated genes in intact rat liver. Sex-specic binding at low- Genet. 1, e16.
but not high-afnity STAT5 sites. Mol. Endocrinol. 23, 12421254. Picardi, A., DAvola, D., Gentilucci, U.V., Galati, G., Fiori, E., Spataro, S., Afeltra, A.,
Lee, T.K., Man, K., Poon, R.T., Lo, C.M., Yuen, A.P., Ng, I.O., Ng, K.T., Leonard, W., Fan, 2006. Diabetes in chronic liver disease: from old concepts to new evidence.
S.T., 2006. Signal transducers and activators of transcription 5b activation Diabetes Metab. Res. Rev. 22, 274283.
enhances hepatocellular carcinoma aggressiveness through induction of Rauch, A., Seitz, S., Baschant, U., Schilling, A.F., Illing, A., Stride, B., Kirilov, M.,
epithelialmesenchymal transition. Cancer Res. 66, 99489956. Mandic, V., Takacz, A., Schmidt-Ullrich, R., Ostermay, S., Schinke, T., Spanbroek,
Lemke, U., Krones-Herzig, A., Berriel Diaz, M., Narvekar, P., Ziegler, A., Vegiopoulos, R., Zaiss, M.M., Angel, P.E., Lerner, U.H., David, J.P., Reichardt, H.M., Amling, M.,
A., Cato, A.C., Bohl, S., Klingmuller, U., Screaton, R.A., Muller-Decker, K., Kersten, Schutz, G., Tuckermann, J.P., 2010. Glucocorticoids suppress bone formation by
S., Herzig, S., 2008. The glucocorticoid receptor controls hepatic dyslipidemia attenuating osteoblast differentiation via the monomeric glucocorticoid
through Hes1. Cell Metab. 8, 212223. receptor. Cell Metab. 11, 517531.
Liang, Y., Osborne, M.C., Monia, B.P., Bhanot, S., Watts, L.M., She, P., DeCarlo, S.O., Reichardt, H.M., Kaestner, K.H., Tuckermann, J., Kretz, O., Wessely, O., Bock, R., Gass,
Chen, X., Demarest, K., 2005. Antisense oligonucleotides targeted against P., Schmid, W., Herrlich, P., Angel, P., Schutz, G., 1998. DNA binding of the
glucocorticoid receptor reduce hepatic glucose production and ameliorate glucocorticoid receptor is not essential for survival. Cell 93, 531541.
hyperglycemia in diabetic mice. Metabolism 54, 848855. Reichardt, H.M., Tuckermann, J.P., Gottlicher, M., Vujic, M., Weih, F., Angel, P.,
Liu, X., Robinson, G.W., Wagner, K.U., Garrett, L., Wynshaw-Boris, A., Hennighausen, Herrlich, P., Schutz, G., 2001. Repression of inammatory responses in the
L., 1997. Stat5a is mandatory for adult mammary gland development and absence of DNA binding by the glucocorticoid receptor. Embo J. 20, 71687173.
lactogenesis. Genes Devlop. 11, 179186. Rogoff, D., Ryder, J.W., Black, K., Yan, Z., Burgess, S.C., McMillan, D.R., White, P.C.,
Liu, Y., Yan, C., Wang, Y., Nakagawa, Y., Nerio, N., Anghel, A., Lutfy, K., Friedman, T.C., 2007. Abnormalities of glucose homeostasis and the hypothalamicpituitary
2006. Liver X receptor agonist T0901317 inhibition of glucocorticoid receptor adrenal axis in mice lacking hexose-6-phosphate dehydrogenase.
expression in hepatocytes may contribute to the amelioration of diabetic Endocrinology 148, 50725080.
syndrome in db/db mice. Endocrinology 147, 50615068. Rosenfeld, R.G., Belgorosky, A., Camacho-Hubner, C., Savage, M.O., Wit, J.M., Hwa, V.,
Liu, Y., Nakagawa, Y., Wang, Y., Liu, L., Du, H., Wang, W., Ren, X., Lutfy, K., Friedman, 2007. Defects in growth hormone receptor signaling. Trends Endocrinol. Metab.
T.C., 2008. Reduction of hepatic glucocorticoid receptor and hexose-6- 18, 134141.
phosphate dehydrogenase expression ameliorates diet-induced obesity and Rosner, W., 1990. The functions of corticosteroid-binding globulin and sex
insulin resistance in mice. J. Mol. Endocrinol. 41, 5364. hormone-binding globulin: recent advances. Endocr. Rev. 11, 8091.
Luedde, T., Beraza, N., Kotsikoris, V., van Loo, G., Nenci, A., De Vos, R., Roskams, T., Rowland, J.E., Lichanska, A.M., Kerr, L.M., White, M., dAniello, E.M., Maher, S.L.,
Trautwein, C., Pasparakis, M., 2007. Deletion of NEMO/IKKgamma in liver Brown, R., Teasdale, R.D., Noakes, P.G., Waters, M.J., 2005. In vivo analysis of
parenchymal cells causes steatohepatitis and hepatocellular carcinoma. Cancer growth hormone receptor signaling domains and their associated transcripts.
Cell 11, 119132. Mol. Cell. Biol. 25, 6677.
Mallette, F.A., Gaumont-Leclerc, M.F., Ferbeyre, G., 2007a. The DNA damage Sapolsky, R.M., Romero, L.M., Munck, A.U., 2000. How do glucocorticoids inuence
signaling pathway is a critical mediator of oncogene-induced senescence. stress responses? Integrating permissive, suppressive, stimulatory, and
Genes Devlop. 21, 4348. preparative actions. Endocr. Rev. 21, 5589.
Mallette, F.A., Gaumont-Leclerc, M.F., Huot, G., Ferbeyre, G., 2007b. Myc down- Scacchi, M., Pincelli, A.I., Cavagnini, F., 1999. Growth hormone in obesity. Int. J.
regulation as a mechanism to activate the Rb pathway in STAT5A-induced Obes. Relat. Metab. Disord. 23, 260271.
senescence. J. Biol. Chem. 282, 3493834944. Schacke, H., Docke, W.D., Asadullah, K., 2002. Mechanisms involved in the side
Michael, M.D., Kulkarni, R.N., Postic, C., Previs, S.F., Shulman, G.I., Magnuson, M.A., effects of glucocorticoids. Pharmacol. Ther. 96, 2343.
Kahn, C.R., 2000. Loss of insulin signaling in hepatocytes leads to severe insulin Schirra, H.J., Anderson, C.G., Wilson, W.J., Kerr, L., Craik, D.J., Waters, M.J., Lichanska,
resistance and progressive hepatic dysfunction. Mol. Cell. 6, 8797. A.M., 2008. Altered metabolism of growth hormone receptor mutant mice: a
Moller, N., Jorgensen, J.O., 2009. Effects of growth hormone on glucose, lipid, and combined NMR metabonomics and microarray study. PLoS ONE 3, e2764.
protein metabolism in human subjects. Endocr. Rev. 30, 152177. Schneider, H.J., Pagotto, U., Stalla, G.K., 2003. Central effects of the somatotropic
Moriggl, R., Sexl, V., Kenner, L., Duntsch, C., Stangl, K., Gingras, S., Hoffmeyer, A., system. Eur. J. Endocrinol. 149, 377392.
Bauer, A., Piekorz, R., Wang, D., Bunting, K.D., Wagner, E.F., Sonneck, K., Valent, Shibli-Rahhal, A., Van Beek, M., Schlechte, J.A., 2006. Cushings syndrome. Clin.
P., Ihle, J.N., Beug, H., 2005. Stat5 tetramer formation is associated with Dermatol. 24, 260265.
leukemogenesis. Cancer Cell 7, 8799. Shteyer, E., Liao, Y., Muglia, L.J., Hruz, P.W., Rudnick, D.A., 2004. Disruption of
Mueller, K.M., Kornfeld, J.W., Friedbichler, K., Blaas, L., Egger, G., Esterbauer, H., hepatic adipogenesis is associated with impaired liver regeneration in mice.
Hasselblatt, P., Schlederer, M., Haindl, S., Wagner, K.U., Engblom, D., Hepatology 40, 13221332.
Haemmerle, G., Kratky, D., Sexl, V., Kenner, L., Kozlov, A.V., Terracciano, L., Sornson, M.W., Wu, W., Dasen, J.S., Flynn, S.E., Norman, D.J., OConnell, S.M.,
Zechner, R., Schuetz, G., Casanova, E., Pospisilik, J.A., Heim, M.H., Moriggl, R., Gukovsky, I., Carriere, C., Ryan, A.K., Miller, A.P., Zuo, L., Gleiberman, A.S.,
2011. Impairment of hepatic growth hormone and glucocorticoid receptor Andersen, B., Beamer, W.G., Rosenfeld, M.G., 1996. Pituitary lineage
signaling causes steatosis and hepatocellular carcinoma in mice. Hepatology 54, determination by the Prophet of Pit-1 homeodomain factor defective in Ames
13981409. dwarsm. Nature 384, 327333.
K.M. Mueller et al. / Molecular and Cellular Endocrinology 361 (2012) 111 11

Sos, B.C., Harris, C., Nordstrom, S.M., Tran, J.L., Balazs, M., Caplazi, P., Febbraio, M., Wang, Z., Masternak, M.M., Al-Regaiey, K.A., Bartke, A., 2007. Adipocytokines and
Applegate, M.A., Wagner, K.U., Weiss, E.J., 2011. Abrogation of growth hormone the regulation of lipid metabolism in growth hormone transgenic and calorie-
secretion rescues fatty liver in mice with hepatocyte-specic deletion of JAK2. J. restricted mice. Endocrinology 148, 28452853.
Clin. Invest. Waters, M.J., Hoang, H.N., Fairlie, D.P., Pelekanos, R.A., Brown, R.J., 2006. New
Starley, B.Q., Calcagno, C.J., Harrison, S.A., 2010. Nonalcoholic fatty liver disease and insights into growth hormone action. J. Mol. Endocrinol. 36, 17.
hepatocellular carcinoma: a weighty connection. Hepatology 51, 18201832. Watts, L.M., Manchem, V.P., Leedom, T.A., Rivard, A.L., McKay, R.A., Bao, D.,
Stoecklin, E., Wissler, M., Gouilleux, F., Groner, B., 1996. Functional interactions Neroladakis, T., Monia, B.P., Bodenmiller, D.M., Cao, J.X., Zhang, H.Y., Cox, A.L.,
between Stat5 and the glucocorticoid receptor. Nature 383, 726728. Jacobs, S.J., Michael, M.D., Sloop, K.W., Bhanot, S., 2005. Reduction of hepatic
Stoecklin, E., Wissler, M., Moriggl, R., Groner, B., 1997. Specic DNA binding of Stat5, and adipose tissue glucocorticoid receptor expression with antisense
but not of glucocorticoid receptor, is required for their functional cooperation in oligonucleotides improves hyperglycemia and hyperlipidemia in diabetic
the regulation of gene transcription. Mol. Cell. Biol. 17, 67086716. rodents without causing systemic glucocorticoid antagonism. Diabetes 54,
Takahashi, Y., Iida, K., Takahashi, K., Yoshioka, S., Fukuoka, H., Takeno, R., Imanaka, 18461853.
M., Nishizawa, H., Takahashi, M., Seo, Y., Hayashi, Y., Kondo, T., Okimura, Y., Kaji, Webster, J.I., Tonelli, L., Sternberg, E.M., 2002. Neuroendocrine regulation of
H., Kitazawa, R., Kitazawa, S., Chihara, K., 2007. Growth hormone reverses immunity. Annu. Rev. Immunol. 20, 125163.
nonalcoholic steatohepatitis in a patient with adult growth hormone deciency. Woele, J., Billiard, J., Rotwein, P., 2003a. Acute control of insulin-like growth factor-
Gastroenterology 132, 938943. I gene transcription by growth hormone through Stat5b. J. Biol. Chem. 278,
Targher, G., Bertolini, L., Rodella, S., Zoppini, G., Zenari, L., Falezza, G., 2006. 2269622702.
Associations between liver histology and cortisol secretion in subjects with Woele, J., Chia, D.J., Rotwein, P., 2003b. Mechanisms of growth hormone (GH)
nonalcoholic fatty liver disease. Clin. Endocrinol. (Oxf.) 64, 337341. action. Identication of conserved Stat5 binding sites that mediate GH-induced
Teglund, S., McKay, C., Schuetz, E., van Deursen, J.M., Stravopodis, D., Wang, D., insulin-like growth factor-I gene activation. J. Biol. Chem. 278, 5126151266.
Brown, M., Bodner, S., Grosveld, G., Ihle, J.N., 1998. Stat5a and Stat5b proteins Xu, J., Lloyd, D.J., Hale, C., Stanislaus, S., Chen, M., Sivits, G., Vonderfecht, S., Hecht, R.,
have essential and nonessential, or redundant, roles in cytokine responses. Cell Li, Y.S., Lindberg, R.A., Chen, J.L., Jung, D.Y., Zhang, Z., Ko, H.J., Kim, J.K., Veniant,
93, 841850. M.M., 2009. Fibroblast growth factor 21 reverses hepatic steatosis, increases
Tronche, F., Opherk, C., Moriggl, R., Kellendonk, C., Reimann, A., Schwake, L., energy expenditure, and improves insulin sensitivity in diet-induced obese
Reichardt, H.M., Stangl, K., Gau, D., Hoeich, A., Beug, H., Schmid, W., Schutz, G., mice. Diabetes 58, 250259.
2004. Glucocorticoid receptor function in hepatocytes is essential to promote Yakar, S., Liu, J.L., Stannard, B., Butler, A., Accili, D., Sauer, B., LeRoith, D., 1999.
postnatal body growth. Genes Devlop. 18, 492497. Normal growth and development in the absence of hepatic insulin-like growth
Tsigos, C., Chrousos, G.P., 2002. Hypothalamicpituitaryadrenal axis, factor I. Proc. Natl. Acad. Sci. USA 96, 73247329.
neuroendocrine factors and stress. J. Psychosom. Res. 53, 865871. Yoo, K.H., Baik, M., Hennighausen, L., 2011. Context-specic growth hormone
Udy, G.B., Towers, R.P., Snell, R.G., Wilkins, R.J., Park, S.H., Ram, P.A., Waxman, D.J., signaling through the transcription factor STAT5: implications for the etiology
Davey, H.W., 1997. Requirement of STAT5b for sexual dimorphism of body of hepatosteatosis and hepatocellular carcinoma. Genes Cancer 2, 39.
growth rates and liver gene expression. Proc. Natl. Acad. Sci. USA 94, 7239 Yu, J.H., Zhu, B.M., Wickre, M., Riedlinger, G., Chen, W., Hosui, A., Robinson, G.W.,
7244. Hennighausen, L., 2011. The transcription factors signal transducer and
Valera, A., Rodriguez-Gil, J.E., Yun, J.S., McGrane, M.M., Hanson, R.W., Bosch, F., 1993. activator of transcription 5A (STAT5A) and STAT5B negatively regulate cell
Glucose metabolism in transgenic mice containing a chimeric P-enolpyruvate proliferation through the activation of cyclin-dependent kinase inhibitor 2b
carboxykinase/bovine growth hormone gene. FASEB J. 7, 791800. (Cdkn2b) and Cdkn1a expression. Hepatology 52, 18081818.
van Schaftingen, E., Gerin, I., 2002. The glucose-6-phosphatase system. Biochem. J. Zhou, Y., Xu, B.C., Maheshwari, H.G., He, L., Reed, M., Lozykowski, M., Okada, S.,
362, 513532. Cataldo, L., Coschigamo, K., Wagner, T.E., Baumann, G., Kopchick, J.J., 1997. A
Vegiopoulos, A., Herzig, S., 2007. Glucocorticoids, metabolism and metabolic mammalian model for Laron syndrome produced by targeted disruption of the
diseases. Mol. Cell. Endocrinol. 275, 4361. mouse growth hormone receptor/binding protein gene (the Laron mouse). Proc.
Viau, V., Meaney, M.J., 2004. Testosterone-dependent variations in plasma and Natl. Acad. Sci. USA 94, 1321513220.
intrapituitary corticosteroid binding globulin and stress hypothalamic Zhu, T., Goh, E.L., Graichen, R., Ling, L., Lobie, P.E., 2001. Signal transduction via the
pituitaryadrenal activity in the male rat. J. Endocrinol. 181, 223231. growth hormone receptor. Cell Signal 13, 599616.
Vijayakumar, A., Novosyadlyy, R., Wu, Y., Yakar, S., LeRoith, D., 2010. Biological
effects of growth hormone on carbohydrate and lipid metabolism. Growth
Horm. IGF Res. 20, 17.

Anda mungkin juga menyukai